1
|
Dörje NM, Shvachiy L, Kück F, Outeiro TF, Strenzke N, Beutner D, Setz C. Age-related alterations in efferent medial olivocochlear-outer hair cell and primary auditory ribbon synapses in CBA/J mice. Front Cell Neurosci 2024; 18:1412450. [PMID: 38988659 PMCID: PMC11234844 DOI: 10.3389/fncel.2024.1412450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/23/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Hearing decline stands as the most prevalent single sensory deficit associated with the aging process. Giving compelling evidence suggesting a protective effect associated with the efferent auditory system, the goal of our study was to characterize the age-related changes in the number of efferent medial olivocochlear (MOC) synapses regulating outer hair cell (OHC) activity compared with the number of afferent inner hair cell ribbon synapses in CBA/J mice over their lifespan. Methods Organs of Corti of 3-month-old CBA/J mice were compared with mice aged between 10 and 20 months, grouped at 2-month intervals. For each animal, one ear was used to characterize the synapses between the efferent MOC fibers and the outer hair cells (OHCs), while the contralateral ear was used to analyze the ribbon synapses between inner hair cells (IHCs) and type I afferent nerve fibers of spiral ganglion neurons (SGNs). Each cochlea was separated in apical, middle, and basal turns, respectively. Results The first significant age-related decline in afferent IHC-SGN ribbon synapses was observed in the basal cochlear turn at 14 months, the middle turn at 16 months, and the apical turn at 18 months of age. In contrast, efferent MOC-OHC synapses in CBA/J mice exhibited a less pronounced loss due to aging which only became significant in the basal and middle turns of the cochlea by 20 months of age. Discussion This study illustrates an age-related reduction on efferent MOC innervation of OHCs in CBA/J mice starting at 20 months of age. Our findings indicate that the morphological decline of efferent MOC-OHC synapses due to aging occurs notably later than the decline observed in afferent IHC-SGN ribbon synapses.
Collapse
Affiliation(s)
- Nele Marie Dörje
- University Medical Center Göttingen, Department of Otolaryngology-Head and Neck Surgery, InnerEarLab, Göttingen, Germany
- University Medical Center Göttingen, Institute for Auditory Neuroscience, Göttingen, Germany
| | - Liana Shvachiy
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
- Institute of Physiology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Cardiovascular Centre, University of Lisbon, Lisbon, Portugal
| | - Fabian Kück
- University Medical Center Göttingen, Department of Medical Statistics, Core Facility Medical Biometry and Statistical Bioinformatics, Göttingen, Germany
| | - Tiago F Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicola Strenzke
- University Medical Center Göttingen, Institute for Auditory Neuroscience, Göttingen, Germany
| | - Dirk Beutner
- University Medical Center Göttingen, Department of Otolaryngology-Head and Neck Surgery, InnerEarLab, Göttingen, Germany
| | - Cristian Setz
- University Medical Center Göttingen, Department of Otolaryngology-Head and Neck Surgery, InnerEarLab, Göttingen, Germany
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| |
Collapse
|
2
|
Gan NS, Oziębło D, Skarżyński H, Ołdak M. Monogenic Causes of Low-Frequency Non-Syndromic Hearing Loss. Audiol Neurootol 2023; 28:327-337. [PMID: 37121227 DOI: 10.1159/000529464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/23/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND Low-frequency non-syndromic hearing loss (LFNSHL) is a rare form of hearing loss (HL). It is defined as HL at low frequencies (≤2,000 Hz) resulting in a characteristic ascending audiogram. LFNSHL is usually diagnosed postlingually and is progressive, leading to HL affecting other frequencies as well. Sometimes it occurs with tinnitus. Around half of the diagnosed prelingual HL cases have a genetic cause and it is usually inherited in an autosomal recessive mode. Postlingual HL caused by genetic changes generally has an autosomal dominant pattern of inheritance and its incidence remains unknown. SUMMARY To date, only a handful of genes have been found as causing LFNSHL: well-established WFS1 and, reported in some cases, DIAPH1, MYO7A, TNC, and CCDC50 (respectively, responsible for DFNA6/14/38, DFNA1, DFNA11, DFNA56, and DFNA44). In this review, we set out audiological phenotypes, causative genetic changes, and molecular mechanisms leading to the development of LFNSHL. KEY MESSAGES LFNSHL is most commonly caused by pathogenic variants in the WFS1 gene, but it is also important to consider changes in other HL genes, which may result in similar audiological phenotype.
Collapse
Affiliation(s)
- Nina Sara Gan
- Department of Genetics, Institute of Physiology and Pathology of Hearing, Warsaw, Poland
| | - Dominika Oziębło
- Department of Genetics, Institute of Physiology and Pathology of Hearing, Warsaw, Poland
| | - Henryk Skarżyński
- Oto-Rhino-Laryngology Surgery Clinic, Institute of Physiology and Pathology of Hearing, Warsaw, Poland
| | - Monika Ołdak
- Department of Genetics, Institute of Physiology and Pathology of Hearing, Warsaw, Poland
| |
Collapse
|
3
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
4
|
Leong YC, Di Foggia V, Pramod H, Bitner-Glindzicz M, Patel A, Sowden JC. Molecular pathology of Usher 1B patient-derived retinal organoids at single cell resolution. Stem Cell Reports 2022; 17:2421-2437. [DOI: 10.1016/j.stemcr.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/08/2022] Open
|
5
|
Shao A, Lopez AJ, Chen J, Tham A, Javier S, Quiroz A, Frick S, Levine EM, Lloyd KCK, Leonard BC, Murphy CJ, Glaser TM, Moshiri A. Arap1 loss causes retinal pigment epithelium phagocytic dysfunction and subsequent photoreceptor death. Dis Model Mech 2022; 15:276063. [PMID: 35758026 PMCID: PMC9346516 DOI: 10.1242/dmm.049343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 06/16/2022] [Indexed: 11/20/2022] Open
Abstract
Retinitis pigmentosa (RP), a retinal degenerative disease, is the leading cause of heritable blindness. Previously, we described that Arap1−/− mice develop a similar pattern of photoreceptor degeneration. Arap1 is an Arf-directed GTPase-activating protein shown to modulate actin cytoskeletal dynamics. Curiously, Arap1 expression was detected in Müller glia and retinal pigment epithelium (RPE), but not the photoreceptors themselves. In this study, we generated conditional knockout mice for Müller glia/RPE, Müller glia and RPE via targeting Rlbp1, Glast and Vmd2 promoters, respectively, to drive Cre recombinase expression to knock out Arap1. Vmd2-Cre Arap1tm1c/tm1c and Rlbp1-Cre Arap1tm1c/tm1c mice, but not Glast-Cre Arap1tm1c/tm1c mice, recapitulated the phenotype originally observed in germline Arap1−/− mice. Mass spectrometry analysis of human ARAP1 co-immunoprecipitation identified candidate binding partners of ARAP1, revealing potential interactants involved in phagocytosis, cytoskeletal composition, intracellular trafficking and endocytosis. Quantification of outer segment phagocytosis in vivo demonstrated a clear phagocytic defect in Arap1−/− mice compared to Arap1+/+ controls. We conclude that Arap1 expression in RPE is necessary for photoreceptor survival due to its indispensable function in RPE phagocytosis. This article has an associated First Person interview with the first author of the paper. Summary: We provide evidence that Arap1 expression in retinal pigment epithelium (RPE) is essential for maintaining photoreceptor health due to its indispensable role in RPE phagocytosis.
Collapse
Affiliation(s)
- Andy Shao
- The University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Antonio Jacobo Lopez
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - JiaJia Chen
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - Addy Tham
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - Seanne Javier
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - Alejandra Quiroz
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - Sonia Frick
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| | - Edward M Levine
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, TN, USA
| | - K C Kent Lloyd
- Mouse Biology Program, U.C. Davis, Davis, CA, USA.,Department of Surgery, School of Medicine, U.C. Davis, Sacramento, CA, USA
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, U.C. Davis, Davis, CA, USA
| | - Christopher J Murphy
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA.,Department of Surgical and Radiological Sciences, School of Veterinary Medicine, U.C. Davis, Davis, CA, USA
| | - Thomas M Glaser
- Department of Cell Biology and Human Anatomy, School of Medicine, U.C. Davis, Davis, CA, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, U.C. Davis, USA
| |
Collapse
|
6
|
Joo SY, Na G, Kim JA, Yoo JE, Kim DH, Kim SJ, Jang SH, Yu S, Kim HY, Choi JY, Gee HY, Jung J. Clinical Heterogeneity Associated with MYO7A Variants Relies on Affected Domains. Biomedicines 2022; 10:biomedicines10040798. [PMID: 35453549 PMCID: PMC9028242 DOI: 10.3390/biomedicines10040798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Autosomal dominant hearing loss (ADHL) manifests as an adult-onset disease or a progressive disease. MYO7A variants are associated with DFNA11, a subtype of ADHL. Here, we examined the role and genotype–phenotype correlation of MYO7A in ADHL. Enrolled families suspected of having post-lingual sensorineural hearing loss were selected for exome sequencing. Mutational alleles in MYO7A were identified according to ACMG guidelines. Segregation analysis was performed to examine whether pathogenic variants segregated with affected status of families. All identified pathogenic variants were evaluated for a phenotype–genotype correlation. MYO7A variants were detected in 4.7% of post-lingual families, and 12 of 14 families were multiplex. Five potentially pathogenic missense variants were identified. Fourteen variants causing autosomal dominant deafness were clustered in motor and MyTH4 domains of MYO7A protein. Missense variants in the motor domain caused late onset of hearing loss with ascending tendency. A severe audiological phenotype was apparent in individuals carrying tail domain variants. We report two new pathogenic variants responsible for DFNA11 in the Korean ADHL population. Dominant pathogenic variants of MYO7A occur frequently in motor and MyTH4 domains. Audiological differences among individuals correspond to specific domains which contain the variants. Therefore, appropriate rehabilitation is needed, particularly for patients with late-onset familial hearing loss.
Collapse
Affiliation(s)
- Sun Young Joo
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Gina Na
- Department of Otorhinolaryngology, Ilsan Paik Hospital, Inje University College of Medicine, Goyang 10380, Korea;
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.E.Y.); (D.H.K.); (J.Y.C.)
| | - Jung Ah Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Jee Eun Yoo
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.E.Y.); (D.H.K.); (J.Y.C.)
| | - Da Hye Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.E.Y.); (D.H.K.); (J.Y.C.)
| | - Se Jin Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Seung Hyun Jang
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Seyoung Yu
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Hye-Youn Kim
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
| | - Jae Young Choi
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.E.Y.); (D.H.K.); (J.Y.C.)
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea; (S.Y.J.); (J.A.K.); (S.J.K.); (S.H.J.); (S.Y.); (H.-Y.K.)
- Correspondence: (H.Y.G.); (J.J.); Tel.: +82-2-2228-0755 (H.Y.G.); +82-2228-3622 (J.J.)
| | - Jinsei Jung
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.E.Y.); (D.H.K.); (J.Y.C.)
- Correspondence: (H.Y.G.); (J.J.); Tel.: +82-2-2228-0755 (H.Y.G.); +82-2228-3622 (J.J.)
| |
Collapse
|
7
|
Johnson Chacko L, Sergi C, Eberharter T, Dudas J, Rask-Andersen H, Hoermann R, Fritsch H, Fischer N, Glueckert R, Schrott-Fischer A. Early appearance of key transcription factors influence the spatiotemporal development of the human inner ear. Cell Tissue Res 2019; 379:459-471. [PMID: 31788757 DOI: 10.1007/s00441-019-03115-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Expression patterns of transcription factors leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5), transforming growth factor-β-activated kinase-1 (TAK1), SRY (sex-determining region Y)-box 2 (SOX2), and GATA binding protein 3 (GATA3) in the developing human fetal inner ear were studied between the gestation weeks 9 and 12. Further development of cochlear apex between gestational weeks 11 and 16 (GW11 and GW16) was examined using transmission electron microscopy. LGR5 was evident in the apical poles of the sensory epithelium of the cochlear duct and the vestibular end organs at GW11. Immunostaining was limited to hair cells of the organ of Corti by GW12. TAK1 was immune positive in inner hair cells of the organ of Corti by GW12 and colocalized with p75 neurotrophic receptor expression. Expression for SOX2 was confined primarily to the supporting cells of utricle at the earliest stage examined at GW9. Intense expression for GATA3 was presented in the cochlear sensory epithelium and spiral ganglia at GW9. Expression of GATA3 was present along the midline of both the utricle and saccule in the zone corresponding to the striolar reversal zone where the hair cell phenotype switches from type I to type II. The spatiotemporal gradient of the development of the organ of Corti was also evident with the apex of the cochlea forming by GW16. It seems that highly specific staining patterns of several transcriptions factors are critical in guiding the genesis of the inner ear over development. Our findings suggest that the spatiotemporal gradient in cochlear development extends at least until gestational week 16.
Collapse
Affiliation(s)
- Lejo Johnson Chacko
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology and Department of Pediatrics, University of Alberta, 8440 112 St, NW, Edmonton, AB, T6G 2B7, Canada
| | - Theresa Eberharter
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Jozsef Dudas
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Helge Rask-Andersen
- Department of Surgical Sciences, Head and Neck Surgery, Section of Otolaryngology, Uppsala University Hospital, SE-751 85, Uppsala, Sweden
| | - Romed Hoermann
- Department of Anatomy, Histology & Embryology, Division of Clinical & Functional Anatomy, Medical University of Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria
| | - Helga Fritsch
- Department of Anatomy, Histology & Embryology, Division of Clinical & Functional Anatomy, Medical University of Innsbruck, Muellerstrasse 59, 6020, Innsbruck, Austria
| | - Natalie Fischer
- University Clinics Innsbruck, Tirol Kliniken, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Rudolf Glueckert
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- University Clinics Innsbruck, Tirol Kliniken, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Anneliese Schrott-Fischer
- Department of Otorhinolaryngology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
8
|
Liu Y, Wei X, Guan L, Xu S, Yuan Y, Lv D, He X, Zhan J, Kong Y, Guo J, Zhang H. Unconventional myosin VIIA promotes melanoma progression. J Cell Sci 2018; 131:jcs.209924. [PMID: 29361540 DOI: 10.1242/jcs.209924] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/11/2018] [Indexed: 11/20/2022] Open
Abstract
Unconventional myosin VIIA (Myo7a) is an actin-based motor molecule that normally functions in the cochlear hair cells of the inner ear. Mutations of MYO7A/Myo7a have been implicated in inherited deafness in both humans and mice. However, there is limited information about the functions of Myo7a outside of the specialized cells of the ears. Herein, we report a previously unidentified function of Myo7a by demonstrating that it plays an important role in melanoma progression. We found that silencing Myo7a by means of RNAi inhibited melanoma cell growth through upregulation of cell cycle regulator p21 (also known as CDKN1A) and suppressed melanoma cell migration and invasion through downregulation of RhoGDI2 (also known as ARHGDIB) and MMP9. Furthermore, Myo7a depletion suppressed melanoma cell metastases to the lung, kidney and bone in mice. In contrast, overexpression of Myo7a promoted melanoma xenograft growth and lung metastasis. Importantly, Myo7a levels are remarkably elevated in human melanoma patients. Collectively, we demonstrated for the first time that Myo7a is able to function in non-specialized cells, a finding that reveals the complicated disease-related roles of Myo7a, especially in melanomas.
Collapse
Affiliation(s)
- Yuqing Liu
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China.,Department of Molecular Neuropathology, Beijing Neurosurgical Institute and Chinese Glioma Cooperative Group (CGCG), Capital Medical University, Beijing 100050, China
| | - Xiaofan Wei
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Lizhao Guan
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Sidi Xu
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Yang Yuan
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Danyu Lv
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Xiaokun He
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Jun Zhan
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, People's Republic of China
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, People's Republic of China
| | - Hongquan Zhang
- Laboratory of Molecular Cell Biology and Tumor Biology, Department of Anatomy, Histology and Embryology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
9
|
Johnson Chacko L, Blumer MJF, Pechriggl E, Rask-Andersen H, Dietl W, Haim A, Fritsch H, Glueckert R, Dudas J, Schrott-Fischer A. Role of BDNF and neurotrophic receptors in human inner ear development. Cell Tissue Res 2017; 370:347-363. [DOI: 10.1007/s00441-017-2686-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/25/2017] [Indexed: 01/03/2023]
|
10
|
Morgan CP, Krey JF, Grati M, Zhao B, Fallen S, Kannan-Sundhari A, Liu XZ, Choi D, Müller U, Barr-Gillespie PG. PDZD7-MYO7A complex identified in enriched stereocilia membranes. eLife 2016; 5:e18312. [PMID: 27525485 PMCID: PMC5005036 DOI: 10.7554/elife.18312] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 08/14/2016] [Indexed: 12/15/2022] Open
Abstract
While more than 70 genes have been linked to deafness, most of which are expressed in mechanosensory hair cells of the inner ear, a challenge has been to link these genes into molecular pathways. One example is Myo7a (myosin VIIA), in which deafness mutations affect the development and function of the mechanically sensitive stereocilia of hair cells. We describe here a procedure for the isolation of low-abundance protein complexes from stereocilia membrane fractions. Using this procedure, combined with identification and quantitation of proteins with mass spectrometry, we demonstrate that MYO7A forms a complex with PDZD7, a paralog of USH1C and DFNB31. MYO7A and PDZD7 interact in tissue-culture cells, and co-localize to the ankle-link region of stereocilia in wild-type but not Myo7a mutant mice. Our data thus describe a new paradigm for the interrogation of low-abundance protein complexes in hair cell stereocilia and establish an unanticipated link between MYO7A and PDZD7.
Collapse
Affiliation(s)
- Clive P Morgan
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, United States
| | - Jocelyn F Krey
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, United States
| | - M'hamed Grati
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, United States
| | - Bo Zhao
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States
| | - Shannon Fallen
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, United States
| | | | - Xue Zhong Liu
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, United States
| | - Dongseok Choi
- OHSU-PSU School of Public Health, Oregon Health and Science University, Portland, United States
- Graduate School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Ulrich Müller
- Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, United States
| | - Peter G Barr-Gillespie
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, United States
| |
Collapse
|
11
|
Li T, Giagtzoglou N, Eberl DF, Jaiswal SN, Cai T, Godt D, Groves AK, Bellen HJ. The E3 ligase Ubr3 regulates Usher syndrome and MYH9 disorder proteins in the auditory organs of Drosophila and mammals. eLife 2016; 5. [PMID: 27331610 PMCID: PMC4978524 DOI: 10.7554/elife.15258] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/21/2016] [Indexed: 01/08/2023] Open
Abstract
Myosins play essential roles in the development and function of auditory organs and multiple myosin genes are associated with hereditary forms of deafness. Using a forward genetic screen in Drosophila, we identified an E3 ligase, Ubr3, as an essential gene for auditory organ development. Ubr3 negatively regulates the mono-ubiquitination of non-muscle Myosin II, a protein associated with hearing loss in humans. The mono-ubiquitination of Myosin II promotes its physical interaction with Myosin VIIa, a protein responsible for Usher syndrome type IB. We show that ubr3 mutants phenocopy pathogenic variants of Myosin II and that Ubr3 interacts genetically and physically with three Usher syndrome proteins. The interactions between Myosin VIIa and Myosin IIa are conserved in the mammalian cochlea and in human retinal pigment epithelium cells. Our work reveals a novel mechanism that regulates protein complexes affected in two forms of syndromic deafness and suggests a molecular function for Myosin IIa in auditory organs.
Collapse
Affiliation(s)
- Tongchao Li
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States
| | - Nikolaos Giagtzoglou
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Department of Neurology, Baylor College of Medicine, Houston, United States
| | - Daniel F Eberl
- Department of Biology, University of Iowa, Iowa City, United States
| | - Sonal Nagarkar Jaiswal
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Tiantian Cai
- Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Dorothea Godt
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Andrew K Groves
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Hugo J Bellen
- Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|
12
|
Lowery J, Jain N, Kuczmarski ER, Mahammad S, Goldman A, Gelfand VI, Opal P, Goldman RD. Abnormal intermediate filament organization alters mitochondrial motility in giant axonal neuropathy fibroblasts. Mol Biol Cell 2015; 27:608-16. [PMID: 26700320 PMCID: PMC4750921 DOI: 10.1091/mbc.e15-09-0627] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/18/2015] [Indexed: 11/17/2022] Open
Abstract
GAN patient cells have abnormal aggregates of vimentin intermediate filaments, to which mitochondria appear to be tethered. Motility of mitochondria, but not lysosomes, is inhibited in these cells. Transfection with wild-type gigaxonin (the protein mutated in this disease) disperses these aggregates and bundles, and mitochondrial motility returns to normal. Giant axonal neuropathy (GAN) is a rare disease caused by mutations in the GAN gene, which encodes gigaxonin, an E3 ligase adapter that targets intermediate filament (IF) proteins for degradation in numerous cell types, including neurons and fibroblasts. The cellular hallmark of GAN pathology is the formation of large aggregates and bundles of IFs. In this study, we show that both the distribution and motility of mitochondria are altered in GAN fibroblasts and this is attributable to their association with vimentin IF aggregates and bundles. Transient expression of wild-type gigaxonin in GAN fibroblasts reduces the number of IF aggregates and bundles, restoring mitochondrial motility. Conversely, silencing the expression of gigaxonin in control fibroblasts leads to changes in IF organization similar to that of GAN patient fibroblasts and a coincident loss of mitochondrial motility. The inhibition of mitochondrial motility in GAN fibroblasts is not due to a global inhibition of organelle translocation, as lysosome motility is normal. Our findings demonstrate that it is the pathological changes in IF organization that cause the loss of mitochondrial motility.
Collapse
Affiliation(s)
- Jason Lowery
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Nikhil Jain
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611 Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Edward R Kuczmarski
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Saleemulla Mahammad
- Stem Cell and Cancer Research Institute, Michael DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anne Goldman
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Puneet Opal
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| |
Collapse
|
13
|
Jordan PM, Fettis M, Holt JC. Efferent innervation of turtle semicircular canal cristae: comparisons with bird and mouse. J Comp Neurol 2015; 523:1258-80. [PMID: 25560461 DOI: 10.1002/cne.23738] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 11/07/2022]
Abstract
In the vestibular periphery of nearly every vertebrate, cholinergic vestibular efferent neurons give rise to numerous presynaptic varicosities that target hair cells and afferent processes in the sensory neuroepithelium. Although pharmacological studies have described the postsynaptic actions of vestibular efferent stimulation in several species, characterization of efferent innervation patterns and the relative distribution of efferent varicosities among hair cells and afferents are also integral to understanding how efferent synapses operate. Vestibular efferent markers, however, have not been well characterized in the turtle, one of the animal models used by our laboratory. Here we sought to identify reliable efferent neuronal markers in the vestibular periphery of turtle, to use these markers to understand how efferent synapses are organized, and to compare efferent neuronal labeling patterns in turtle with two other amniotes using some of the same markers. Efferent fibers and varicosities were visualized in the semicircular canal of red-eared turtles (Trachemys scripta elegans), zebra finches (Taeniopygia guttata), and mice (Mus musculus) utilizing fluorescent immunohistochemistry with antibodies against choline acetyltransferase (ChAT). Vestibular hair cells and afferents were counterstained using antibodies to myosin VIIa and calretinin. In all species, ChAT labeled a population of small diameter fibers giving rise to numerous spherical varicosities abutting type II hair cells and afferent processes. That these ChAT-positive varicosities represent presynaptic release sites were demonstrated by colabeling with antibodies against the synaptic vesicle proteins synapsin I, SV2, or syntaxin and the neuropeptide calcitonin gene-related peptide. Comparisons of efferent innervation patterns among the three species are discussed.
Collapse
Affiliation(s)
- Paivi M Jordan
- Department of Otolaryngology, University of Rochester, Rochester, New York
| | | | | |
Collapse
|
14
|
Aboul Naga SH, Dithmer M, Chitadze G, Kabelitz D, Lucius R, Roider J, Klettner A. Intracellular pathways following uptake of bevacizumab in RPE cells. Exp Eye Res 2015; 131:29-41. [DOI: 10.1016/j.exer.2014.12.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 12/11/2014] [Accepted: 12/17/2014] [Indexed: 12/23/2022]
|
15
|
Ogun O, Zallocchi M. Clarin-1 acts as a modulator of mechanotransduction activity and presynaptic ribbon assembly. ACTA ACUST UNITED AC 2014; 207:375-91. [PMID: 25365995 PMCID: PMC4226736 DOI: 10.1083/jcb.201404016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Clarin-1 is a four-transmembrane protein expressed by hair cells and photoreceptors. Mutations in its corresponding gene are associated with Usher syndrome type 3, characterized by late-onset and progressive hearing and vision loss in humans. Mice carrying mutations in the clarin-1 gene have hair bundle dysmorphology and a delay in synapse maturation. In this paper, we examined the expression and function of clarin-1 in zebrafish hair cells. We observed protein expression as early as 1 d postfertilization. Knockdown of clarin-1 resulted in inhibition of FM1-43 incorporation, shortening of the kinocilia, and mislocalization of ribeye b clusters. These phenotypes were fully prevented by co-injection with clarin-1 transcript, requiring its C-terminal tail. We also observed an in vivo interaction between clarin-1 and Pcdh15a. Altogether, our results suggest that clarin-1 is functionally important for mechanotransduction channel activity and for proper localization of synaptic components, establishing a critical role for clarin-1 at the apical and basal poles of hair cells.
Collapse
Affiliation(s)
- Oluwatobi Ogun
- Sensory Neuroscience Department, Boys Town National Research Hospital, Omaha, NE 68131
| | - Marisa Zallocchi
- Sensory Neuroscience Department, Boys Town National Research Hospital, Omaha, NE 68131
| |
Collapse
|
16
|
Pujol R, Pickett SB, Nguyen TB, Stone JS. Large basolateral processes on type II hair cells are novel processing units in mammalian vestibular organs. J Comp Neurol 2014; 522:3141-59. [PMID: 24825750 DOI: 10.1002/cne.23625] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 05/05/2014] [Accepted: 05/06/2014] [Indexed: 12/29/2022]
Abstract
Sensory receptors in the vestibular system (hair cells) encode head movements and drive central motor reflexes that control gaze, body movements, and body orientation. In mammals, type I and II vestibular hair cells are defined by their shape, contacts with vestibular afferent nerves, and membrane conductance. Here we describe unique morphological features of type II vestibular hair cells in mature rodents (mice and gerbils) and bats. These features are cytoplasmic processes that extend laterally from the hair cell base and project under type I hair cells. Closer analysis of adult mouse utricles demonstrated that the basolateral processes of type II hair cells vary in shape, size, and branching, with the longest processes extending three to four hair cell widths. The hair cell basolateral processes synapse upon vestibular afferent nerves and receive inputs from vestibular efferent nerves. Furthermore, some basolateral processes make physical contacts with the processes of other type II hair cells, forming some sort of network among type II hair cells. Basolateral processes are rare in perinatal mice and do not attain their mature form until 3-6 weeks of age. These observations demonstrate that basolateral processes are significant signaling regions of type II vestibular hair cells and suggest that type II hair cells may directly communicate with each other, which has not been described in vertebrates.
Collapse
Affiliation(s)
- Rémy Pujol
- The Virginia Merrill Bloedel Hearing Research Center, and the Department of Otolaryngology-Head and Neck Surgery, University of Washington, Seattle, Washington, 98195-7923; INSERM Unit 1051, Institute of Neuroscience, 34091, Montpellier, France
| | | | | | | |
Collapse
|
17
|
Zallocchi M, Binley K, Lad Y, Ellis S, Widdowson P, Iqball S, Scripps V, Kelleher M, Loader J, Miskin J, Peng YW, Wang WM, Cheung L, Delimont D, Mitrophanous KA, Cosgrove D. EIAV-based retinal gene therapy in the shaker1 mouse model for usher syndrome type 1B: development of UshStat. PLoS One 2014; 9:e94272. [PMID: 24705452 PMCID: PMC3976400 DOI: 10.1371/journal.pone.0094272] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 03/14/2014] [Indexed: 01/21/2023] Open
Abstract
Usher syndrome type 1B is a combined deaf-blindness condition caused by mutations in the MYO7A gene. Loss of functional myosin VIIa in the retinal pigment epithelia (RPE) and/or photoreceptors leads to blindness. We evaluated the impact of subretinally delivered UshStat, a recombinant EIAV-based lentiviral vector expressing human MYO7A, on photoreceptor function in the shaker1 mouse model for Usher type 1B that lacks a functional Myo7A gene. Subretinal injections of EIAV-CMV-GFP, EIAV-RK-GFP (photoreceptor specific), EIAV-CMV-MYO7A (UshStat) or EIAV-CMV-Null (control) vectors were performed in shaker1 mice. GFP and myosin VIIa expression was evaluated histologically. Photoreceptor function in EIAV-CMV-MYO7A treated eyes was determined by evaluating α-transducin translocation in photoreceptors in response to low light intensity levels, and protection from light induced photoreceptor degeneration was measured. The safety and tolerability of subretinally delivered UshStat was evaluated in macaques. Expression of GFP and myosin VIIa was confirmed in the RPE and photoreceptors in shaker1 mice following subretinal delivery of the EIAV-CMV-GFP/MYO7A vectors. The EIAV-CMV-MYO7A vector protected the shaker1 mouse photoreceptors from acute and chronic intensity light damage, indicated by a significant reduction in photoreceptor cell loss, and restoration of the α-transducin translocation threshold in the photoreceptors. Safety studies in the macaques demonstrated that subretinal delivery of UshStat is safe and well-tolerated. Subretinal delivery of EIAV-CMV-MYO7A (UshStat) rescues photoreceptor phenotypes in the shaker1 mouse. In addition, subretinally delivered UshStat is safe and well-tolerated in macaque safety studies These data support the clinical development of UshStat to treat Usher type 1B syndrome.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Katie Binley
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Yatish Lad
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Scott Ellis
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Peter Widdowson
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Sharifah Iqball
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Vicky Scripps
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Michelle Kelleher
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - Julie Loader
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - James Miskin
- Oxford BioMedica (UK) Ltd, Oxford Science Park, Oxford, United Kingdom
| | - You-Wei Peng
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Wei-Min Wang
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Linda Cheung
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Duane Delimont
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | | | - Dominic Cosgrove
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
18
|
Bonilha VL. Retinal pigment epithelium (RPE) cytoskeleton in vivo and in vitro. Exp Eye Res 2013; 126:38-45. [PMID: 24090540 DOI: 10.1016/j.exer.2013.09.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/18/2013] [Accepted: 09/19/2013] [Indexed: 12/31/2022]
Abstract
The retinal pigment epithelium (RPE) constitutes a monolayer of cuboidal cells that interact apically with the interphotoreceptor matrix (IPM) and outer segments of the photoreceptor cells and basally with the subjacent Bruch's membrane. This highly polarized structure is maintained by the cytoskeleton of individual cells and their interactions at the basolateral junctional complexes that stabilize this epithelial structure. This RPE complex network of filaments, tubules and associated proteins is modeled by the cellular environment, the RPE intercellular interactions, and by its interactions with the extracellular matrix. This is a review of the key features of the RPE cytoskeleton in vivo and in vitro.
Collapse
Affiliation(s)
- Vera L Bonilha
- Department of Ophthalmic Research, The Cole Eye Institute, Cleveland Clinic Lerner College of Medicine, 9500 Euclid Avenue i31, Cleveland, OH 44195, USA.
| |
Collapse
|
19
|
Müller M, Diensthuber RP, Chizhov I, Claus P, Heissler SM, Preller M, Taft MH, Manstein DJ. Distinct functional interactions between actin isoforms and nonsarcomeric myosins. PLoS One 2013; 8:e70636. [PMID: 23923011 PMCID: PMC3724804 DOI: 10.1371/journal.pone.0070636] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 06/26/2013] [Indexed: 02/07/2023] Open
Abstract
Despite their near sequence identity, actin isoforms cannot completely replace each other in vivo and show marked differences in their tissue-specific and subcellular localization. Little is known about isoform-specific differences in their interactions with myosin motors and other actin-binding proteins. Mammalian cytoplasmic β- and γ-actin interact with nonsarcomeric conventional myosins such as the members of the nonmuscle myosin-2 family and myosin-7A. These interactions support a wide range of cellular processes including cytokinesis, maintenance of cell polarity, cell adhesion, migration, and mechano-electrical transduction. To elucidate differences in the ability of isoactins to bind and stimulate the enzymatic activity of individual myosin isoforms, we characterized the interactions of human skeletal muscle α-actin, cytoplasmic β-actin, and cytoplasmic γ-actin with human myosin-7A and nonmuscle myosins-2A, -2B and -2C1. In the case of nonmuscle myosins-2A and -2B, the interaction with either cytoplasmic actin isoform results in 4-fold greater stimulation of myosin ATPase activity than was observed in the presence of α-skeletal muscle actin. Nonmuscle myosin-2C1 is most potently activated by β-actin and myosin-7A by γ-actin. Our results indicate that β- and γ-actin isoforms contribute to the modulation of nonmuscle myosin-2 and myosin-7A activity and thereby to the spatial and temporal regulation of cytoskeletal dynamics. FRET-based analyses show efficient copolymerization abilities for the actin isoforms in vitro. Experiments with hybrid actin filaments show that the extent of actomyosin coupling efficiency can be regulated by the isoform composition of actin filaments.
Collapse
Affiliation(s)
- Mirco Müller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Igor Chizhov
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Peter Claus
- Institute of Neuroanatomy, Hannover Medical School, Hannover, Germany
| | - Sarah M. Heissler
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Manuel H. Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Dietmar J. Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
20
|
Ladrech S, Mathieu M, Puel JL, Lenoir M. Supporting cells regulate the remodelling of aminoglycoside-injured organ of Corti, through the release of high mobility group box 1. Eur J Neurosci 2013; 38:2962-72. [DOI: 10.1111/ejn.12290] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 11/28/2022]
|
21
|
Liu F, Li P, Liu Y, Li W, Wong F, Du R, Wang L, Li C, Jiang F, Tang Z, Liu M. Novel compound heterozygous mutations in MYO7A in a Chinese family with Usher syndrome type 1. Mol Vis 2013; 19:695-701. [PMID: 23559863 PMCID: PMC3611938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 03/19/2013] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To identify the disease-causing mutation(s) in a Chinese family with autosomal recessive Usher syndrome type 1 (USH1). METHODS An ophthalmic examination and an audiometric test were conducted to ascertain the phenotype of two affected siblings. The microsatellite marker D11S937, which is close to the candidate gene MYO7A (USH1B locus), was selected for genotyping. From the DNA of the proband, all coding exons and exon-intron boundaries of MYO7A were sequenced to identify the disease-causing mutation(s). Restriction fragment length polymorphism (RFLP) analysis was performed to exclude the alternative conclusion that the mutations are non-pathogenic rare polymorphisms. RESULTS Based on severe hearing impairment, unintelligible speech, and retinitis pigmentosa, a clinical diagnosis of Usher syndrome type 1 was made. The genotyping results did not exclude the USH1B locus, which suggested that the MYO7A gene was likely the gene associated with the disease-causing mutation(s) in the family. With direct DNA sequencing of MYO7A, two novel compound heterozygous mutations (c.3742G>A and c.6051+1G>A) of MYO7A were identified in the proband. DNA sequence analysis and RFLP analysis of other family members showed that the mutations cosegregated with the disease. Unaffected members, including the parents, uncle, and sister of the proband, carry only one of the two mutations. The mutations were not present in the controls (100 normal Chinese subjects=200 chromosomes) according to the RFLP analysis. CONCLUSIONS In this study, we identified two novel mutations, c.3742G>A (p.E1248K) and c.6051+1G>A (donor splice site mutation in intron 44), of MYO7A in a Chinese non-consanguineous family with USH1. The mutations cosegregated with the disease and most likely cause the phenotype in the two affected siblings who carry these mutations compound heterozygously. Our finding expands the mutational spectrum of MYO7A.
Collapse
Affiliation(s)
- Fei Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Li
- The Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Ying Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Weirong Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Fulton Wong
- Departments of Ophthalmology and Neurobiology, Duke University School of Medicine, Durham, NC
| | - Rong Du
- The Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Lei Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Chang Li
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Fagang Jiang
- The Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Zhaohui Tang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Affiliation(s)
- M Amanda Hartman
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
23
|
Retinal gene therapy with a large MYO7A cDNA using adeno-associated virus. Gene Ther 2013; 20:824-33. [PMID: 23344065 PMCID: PMC3640772 DOI: 10.1038/gt.2013.3] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 11/12/2012] [Accepted: 12/27/2012] [Indexed: 11/11/2022]
Abstract
Usher 1 patients are born profoundly deaf and then develop retinal degeneration. Thus they are readily identified prior to the onset of retinal degeneration, making gene therapy a viable strategy to prevent their blindness. Here, we have investigated the use of adeno-associated viruses (AAV) for the delivery of the Usher 1B gene, MYO7A, to retinal cells in cell culture and in Myo7a-null mice. MYO7A cDNA, under control of a smCBA promoter, was packaged in single AAV2 and AAV5 vectors, and as two overlapping halves in dual AAV2 vectors. The 7.9-kb smCBA-MYO7A exceeds the capacity of an AAV vector; packaging of such oversized constructs into single AAV vectors may involve fragmentation of the gene. Nevertheless, the AAV2 and AAV5 single vector preparations successfully transduced photoreceptor and RPE cells, resulting in functional, full-length MYO7A protein and correction of mutant phenotypes, suggesting successful homologous recombination of gene fragments. With discrete, conventional-sized dual AAV2 vectors, full-length MYO7A was detected, but the level of protein expression was variable, and only a minority of cells showed phenotype correction. Our results show that MYO7A therapy with AAV2 or AAV5 single vectors is efficacious, however, the dual AAV2 approach proved to be less effective.
Collapse
|
24
|
Zallocchi M, Meehan DT, Delimont D, Rutledge J, Gratton MA, Flannery J, Cosgrove D. Role for a novel Usher protein complex in hair cell synaptic maturation. PLoS One 2012; 7:e30573. [PMID: 22363448 PMCID: PMC3281840 DOI: 10.1371/journal.pone.0030573] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 12/22/2011] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1−/− mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzerav3J mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Daniel T. Meehan
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Duane Delimont
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | - Joseph Rutledge
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - Michael Anne Gratton
- Otolaryngology-Head, Neck Surgery, St Louis University, St Louis, Missouri, United States of America
| | - John Flannery
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Dominic Cosgrove
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
25
|
Abstract
Mutations in MYO7A (myosin VIIa) cause Usher syndrome type 1B, a disorder involving profound congenital deafness and progressive blindness. In the retina, most MYO7A is localized in the apical region of the RPE (retinal pigmented epithelial) cells, and a small amount is associated with the ciliary and periciliary membranes of the photoreceptor cells. Its roles appear to be quite varied. Studies with MYO7A-null mice indicate that MYO7A participates in the apical localization of RPE melanosomes and in the removal of phagosomes from the apical RPE for their delivery to lysosomes in the basal RPE. In the first role, MYO7A competes with microtubule motors, but in the second one, it may function co-operatively. An additional role of MYO7A in the RPE is indicated by the requirement for it in the light-dependent translocation of the ER (endoplasmic reticulum)-associated visual cycle enzyme RPE65 and normal functioning of the visual retinoid cycle. In photoreceptor cells lacking MYO7A, opsin accumulates abnormally in the transition zone of the cilium, suggesting that MYO7A functions as a selective barrier for membrane proteins at the distal end of the transition zone. It is likely that the progressive retinal degeneration that occurs in Usher syndrome 1B patients results from a combination of cellular defects in the RPE and photoreceptor cells.
Collapse
|
26
|
Zallocchi M, Meehan DT, Delimont D, Rutledge J, Gratton MA, Flannery J, Cosgrove D. Role for a novel Usher protein complex in hair cell synaptic maturation. PLoS One 2012; 7:e30573. [PMID: 22363448 DOI: 10.1371/journal.pone.0030573pone-d-11-06651[pii]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 12/22/2011] [Indexed: 05/19/2023] Open
Abstract
The molecular mechanisms underlying hair cell synaptic maturation are not well understood. Cadherin-23 (CDH23), protocadherin-15 (PCDH15) and the very large G-protein coupled receptor 1 (VLGR1) have been implicated in the development of cochlear hair cell stereocilia, while clarin-1 has been suggested to also play a role in synaptogenesis. Mutations in CDH23, PCDH15, VLGR1 and clarin-1 cause Usher syndrome, characterized by congenital deafness, vestibular dysfunction and retinitis pigmentosa. Here we show developmental expression of these Usher proteins in afferent spiral ganglion neurons and hair cell synapses. We identify a novel synaptic Usher complex comprised of clarin-1 and specific isoforms of CDH23, PCDH15 and VLGR1. To establish the in vivo relevance of this complex, we performed morphological and quantitative analysis of the neuronal fibers and their synapses in the Clrn1-/- mouse, which was generated by incomplete deletion of the gene. These mice showed a delay in neuronal/synaptic maturation by both immunostaining and electron microscopy. Analysis of the ribbon synapses in Ames waltzer(av3J) mice also suggests a delay in hair cell synaptogenesis. Collectively, these results show that, in addition to the well documented role for Usher proteins in stereocilia development, Usher protein complexes comprised of specific protein isoforms likely function in synaptic maturation as well.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Boys Town National Research Hospital, Omaha, Nebraska, United States of America
| | | | | | | | | | | | | |
Collapse
|
27
|
Towers ER, Kelly JJ, Sud R, Gale JE, Dawson SJ. Caprin-1 is a target of the deafness gene Pou4f3 and is recruited to stress granules in cochlear hair cells in response to ototoxic damage. J Cell Sci 2011; 124:1145-55. [PMID: 21402877 DOI: 10.1242/jcs.076141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The POU4 family of transcription factors are required for survival of specific cell types in different sensory systems. Pou4f3 is essential for the survival of auditory sensory hair cells and several mutations in human POU4F3 cause hearing loss. Thus, genes regulated by Pou4f3 are likely to be essential for hair cell survival. We performed a subtractive hybridisation screen in an inner-ear-derived cell line to find genes with differential expression in response to changes in Pou4f3 levels. The screen identified the stress-granule-associated protein Caprin-1 as being downregulated by Pou4f3. We demonstrated that this regulation occurs through the direct interaction of Pou4f3 with binding sites in the Caprin-1 5' flanking sequence, and describe the expression pattern of Caprin-1 mRNA and protein in the cochlea. Moreover, we found Caprin-1-containing stress granules are induced in cochlear hair cells following aminoglycoside-induced damage. This is the first report of stress granule formation in mammalian hair cells and suggests that the formation of Caprin-1-containing stress granules is a key damage response to a clinically relevant ototoxic agent. Our results have implications for the understanding of aminoglycoside-induced hearing loss and provide further evidence that stress granule formation is a fundamental cellular stress response.
Collapse
Affiliation(s)
- Emily R Towers
- UCL Ear Institute, 332 Gray's Inn Road, London WC1X 8EE, UK
| | | | | | | | | |
Collapse
|
28
|
Heissler SM, Manstein DJ. Functional characterization of the human myosin-7a motor domain. Cell Mol Life Sci 2011; 69:299-311. [PMID: 21687988 PMCID: PMC3249170 DOI: 10.1007/s00018-011-0749-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 11/26/2022]
Abstract
Myosin-7a participates in auditory and visual processes. Defects in MYO7A, the gene encoding the myosin-7a heavy chain, are causative for Usher syndrome 1B, the most frequent cause of deaf-blindness in humans. In the present study, we performed a detailed kinetic and functional characterization of the isolated human myosin-7a motor domain to elucidate the details of chemomechanical coupling and the regulation of motor function. A rate-limiting, slow ADP release step causes long lifetimes of strong actin-binding intermediates and results in a high duty ratio. Moreover, our results reveal a Mg2+-sensitive regulatory mechanism tuning the kinetic and mechanical properties of the myosin-7a motor domain. We obtained direct evidence that changes in the concentration of free Mg2+ ions affect the motor properties of human myosin-7a using an in vitro motility assay system. Our results suggest that in a cellular environment, compartment-specific fluctuations in free Mg2+ ions can mediate the conditional switching of myosin-7a between cargo moving and tension bearing modes.
Collapse
Affiliation(s)
- Sarah M. Heissler
- Institut für Biophysikalische Chemie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Dietmar J. Manstein
- Institut für Biophysikalische Chemie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
29
|
Targeting of the hair cell proteins cadherin 23, harmonin, myosin XVa, espin, and prestin in an epithelial cell model. J Neurosci 2010; 30:7187-201. [PMID: 20505086 DOI: 10.1523/jneurosci.0852-10.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We have developed an advantageous epithelial cell transfection model for examining the targeting, interactions, and mutations of hair cell proteins. When expressed in LLC-PK1-CL4 epithelial cells (CL4 cells), the outer hair cell protein prestin showed faithful domain-specific targeting to the basolateral plasma membrane. We examined the consequences of mutations affecting prestin activity and assigned a targeting role to the cytoplasmic tail. The stereociliary link protein cadherin 23 (Cdh23) was targeted to the plasma membrane of CL4 cell microvilli, the topological equivalent of stereocilia. In cells coexpressing the Cdh23 cytoplasmic binding protein harmonin, a large fraction of harmonin became colocalized with Cdh23 in microvilli. Using this assay and in vitro protein binding assays, we formulated an alternative model for Cdh23-harmonin binding, in which the primary interaction is between the harmonin N-domain and a 35-residue internal peptide in the Cdh23 cytoplasmic tail. Contrary to a previous model, we found no role for the Cdh23 C-terminal PDZ (PSD-95/Dlg/ZO-1)-binding motif and observed that Cdh23 bound similar levels of harmonin with or without the exon 68 peptide. We also examined two proteins involved in stereocilium elongation. The stereociliary actin-bundling protein espin was targeted to CL4 cell microvilli and caused microvillar elongation, whereas espin with the c.2469delGTCA or c.1988delAGAG human deafness mutation showed defects in microvillar targeting and elongation. The unconventional myosin motor myosin XVa accumulated at the tips of espin-elongated microvilli, by analogy to its location in stereocilia, whereas myosin XVa with the c.4351G>A or c.4669A>G human deafness mutation did not, revealing functional deficits in motor activity.
Collapse
|
30
|
Zallocchi M, Sisson JH, Cosgrove D. Biochemical characterization of native Usher protein complexes from a vesicular subfraction of tracheal epithelial cells. Biochemistry 2010; 49:1236-47. [PMID: 20058854 DOI: 10.1021/bi9020617] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Usher syndrome is the major cause of deaf/blindness in the world. It is a genetic heterogeneous disorder, with nine genes already identified as causative for the disease. We noted expression of all known Usher proteins in bovine tracheal epithelial cells and exploited this system for large-scale biochemical analysis of Usher protein complexes. The dissected epithelia were homogenized in nondetergent buffer and sedimented on sucrose gradients. At least two complexes were evident after the first gradient: one formed by specific isoforms of CDH23, PCDH15, and VLGR-1 and a different one at the top of the gradient that included all of the Usher proteins and rab5, a transport vesicle marker. TEM analysis of these top fractions found them enriched in 100-200 nm vesicles, confirming a vesicular association of the Usher complex(es). Immunoisolation of these vesicles confirmed some of the associations already predicted and identified novel interactions. When the vesicles are lysed in the presence of phenylbutyrate, most of the Usher proteins cosediment into the gradient at a sedimentation coefficient of approximately 50 S, correlating with a predicted molecular mass of 2 x 10(6) Da. Although it is still unclear whether there is only one complex or several independent complexes that are trafficked within distinct vesicular pools, this work shows for the first time that native Usher protein complexes occur in vivo. This complex(es) is present primarily in transport vesicles at the apical pole of tracheal epithelial cells, predicting that Usher proteins may be directionally transported as complexes in hair cells and photoreceptors.
Collapse
Affiliation(s)
- Marisa Zallocchi
- Usher Syndrome Center, Boys Town National Research Hospital, 555 North 30th Street, Omaha, Nebraska 68131, USA
| | | | | |
Collapse
|
31
|
A novel allele of myosin VIIa reveals a critical function for the C-terminal FERM domain for melanosome transport in retinal pigment epithelial cells. J Neurosci 2010; 29:15810-8. [PMID: 20016096 DOI: 10.1523/jneurosci.4876-09.2009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Mutations in the head and tail domains of the motor protein myosin VIIA (MYO7A) cause deaf-blindness (Usher syndrome type 1B, USH1B) and nonsyndromic deafness (DFNB2, DFNA11). The head domain binds to F-actin and serves as the MYO7A motor domain, but little is known about the function of the tail domain. In a genetic screen, we have identified polka mice, which carry a mutation (c.5742 + 5G > A) that affects splicing of the MYO7A transcript and truncates the MYO7A tail domain at the C-terminal FERM domain. In the inner ear, expression of the truncated MYO7A protein is severely reduced, leading to defects in hair cell development. In retinal pigment epithelial (RPE) cells, the truncated MYO7A protein is expressed at comparative levels to wild-type protein but fails to associate with and transport melanosomes. We conclude that the C-terminal FERM domain of MYO7A is critical for melanosome transport in RPE cells. Our findings also suggest that MYO7A mutations can lead to tissue-specific effects on protein levels, which may explain why some mutations in MYO7A lead to deafness without retinal impairment.
Collapse
|
32
|
Lopes VS, Ramalho JS, Owen DM, Karl MO, Strauss O, Futter CE, Seabra MC. The ternary Rab27a-Myrip-Myosin VIIa complex regulates melanosome motility in the retinal pigment epithelium. Traffic 2008; 8:486-99. [PMID: 17451552 PMCID: PMC1920545 DOI: 10.1111/j.1600-0854.2007.00548.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The retinal pigment epithelium (RPE) contains melanosomes similar to those found in the skin melanocytes, which undergo dramatic light-dependent movements in fish and amphibians. In mammals, those movements are more subtle and appear to be regulated by the Rab27a GTPase and the unconventional myosin, Myosin VIIa (MyoVIIa). Here we address the hypothesis that a recently identified Rab27a- and MyoVIIa-interacting protein, Myrip, promotes the formation of a functional tripartite complex. In heterologous cultured cells, all three proteins co-immunoprecipitated following overexpression. Rab27a and Myrip localize to the peripheral membrane of RPE melanosomes as observed by immunofluorescence and immunoelectron microscopy. Melanosome dynamics were studied using live-cell imaging of mouse RPE primary cultures. Wild-type RPE melanosomes exhibited either stationary or slow movement interrupted by bursts of fast movement, with a peripheral directionality trend. Nocodazole treatment led to melanosome paralysis, suggesting that movement requires microtubule motors. Significant and similar alterations in melanosome dynamics were observed when any one of the three components of the complex was missing, as studied in ashen- (Rab27a defective) and shaker-1 (MyoVIIa mutant)-derived RPE cells, and in wild-type RPE cells transduced with adenovirus carrying specific sequences to knockdown Myrip expression. We observed a significant increase in the number of motile melanosomes, exhibiting more frequent and prolonged bursts of fast movement, and inversion of directionality. Similar alterations were observed upon cytochalasin D treatment, suggesting that the Rab27a–Myrip–MyoVIIa complex regulates tethering of melanosomes onto actin filaments, a process that ensures melanosome movement towards the cell periphery.
Collapse
Affiliation(s)
- Vanda S Lopes
- Molecular and Cellular Medicine, Faculty of Medicine, Imperial College LondonLondon SW7 2AZ, UK
| | - José S Ramalho
- Centre of Ophthalmology, Biomedical Institute for Research in Light and Image, University of Coimbra3000-354 Coimbra, Portugal
| | - Dylan M Owen
- Chemical Biology Centre, Imperial College LondonLondon, SW7 2AZ, UK
| | - Mike O Karl
- Bereich Experimentelle Ophthalmologie, Klinik und Poliklinik fuer Augenheilkunde, Universitaetsklinikum Hamburg-Eppendorf20246, Hamburg, Germany
| | - Olaf Strauss
- Bereich Experimentelle Ophthalmologie, Klinik und Poliklinik fuer Augenheilkunde, Universitaetsklinikum Hamburg-Eppendorf20246, Hamburg, Germany
| | - Clare E Futter
- Division of Cell Biology, Institute of Ophthalmology, University College LondonLondon EC1V 9EL, UK
| | - Miguel C Seabra
- Molecular and Cellular Medicine, Faculty of Medicine, Imperial College LondonLondon SW7 2AZ, UK
- *Corresponding author: Miguel C. Seabra,
| |
Collapse
|
33
|
Klomp AE, Teofilo K, Legacki E, Williams DS. Analysis of the linkage of MYRIP and MYO7A to melanosomes by RAB27A in retinal pigment epithelial cells. ACTA ACUST UNITED AC 2007; 64:474-87. [PMID: 17352418 DOI: 10.1002/cm.20198] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The apical region of the retinal pigment epithelium (RPE) typically contains melanosomes. Their apical distribution is dependent on RAB27A and the unconventional myosin, MYO7A. Evidence from studies using in vitro binding assays, melanocyte transfection, and immunolocalization have indicated that the exophilin, MYRIP, links RAB27A on melanosomes to MYO7A, analogous to the manner that melanophilin links RAB27A on melanocyte melanosomes to MYO5A. To test the functionality of this hypothesis in RPE cells, we have examined the relationship among MYRIP, RAB27A and MYO7A with studies of RPE cells in primary culture (including live-cell imaging), analyses of mutant mouse retinas, and RPE cell fractionation experiments. Our results indicate that the retinal distribution of MYRIP is limited to the RPE, mainly the apical region. In RPE cells, RAB27A, MYRIP, and MYO7A were all associated with melanosomes, undergoing both slow and rapid movements. Analyses of mutant mice provide genetic evidence that MYRIP is linked to melanosomes via RAB27A, but show that recruitment of MYRIP to apical RPE is independent of melanosomes and RAB27A. RAB27A and MYRIP also associated with motile small vesicles of unknown origin. The present results provide evidence from live RPE cells that the RAB27A-MYRIP-MYO7A complex functions in melanosome motility. They also demonstrate that RAB27A provides an essential link to the melanosome.
Collapse
Affiliation(s)
- Adriana E Klomp
- Department of Pharmacology, UCSD School of Medicine, La Jolla, CA 92093-0912, USA
| | | | | | | |
Collapse
|
34
|
Abstract
The actin network has been implicated in the intracellular transport and positioning of the melanosomes, organelles that are specialized in the biosynthesis and the storage of melanin. It contributes also to molecular mechanisms that underlie the intracellular membrane dynamics and thereby can control the biogenesis of melanosomes. Two mechanisms for actin-based movements have been identified: one is dependent on the motors associated to actin namely the myosins; the other is dependent on actin polymerization. This review will focus on to the role of the actin cytoskeleton and myosins in the transport and in the biogenesis of melanosomes. Myosins involved in membrane traffic are largely seen as transporters of organelles or membrane vesicles containing cargos along the actin networks. Yet increasing evidence suggests that some of the myosins contribute to the dynamics of internal membrane by using other mechanisms. The role of the myosins and the different molecular mechanisms by which they contribute or may contribute to the distribution, the movement and the biogenesis of the melanosomes in epidermal melanocytes and retinal pigmented epithelial (RPE) cells will be discussed.
Collapse
|
35
|
O'Connell CB, Tyska MJ, Mooseker MS. Myosin at work: motor adaptations for a variety of cellular functions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1773:615-30. [PMID: 16904206 DOI: 10.1016/j.bbamcr.2006.06.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2006] [Revised: 05/22/2006] [Accepted: 06/30/2006] [Indexed: 12/30/2022]
Abstract
Cells have evolved multiple mechanisms to overcome the effects of entropy and diffusion to create a highly ordered environment. For cells to function properly, some components must be anchored to provide a framework or structure. Others must be rapidly transported over long distances to generate asymmetries in cell morphology and composition. To accomplish long-range transport, cells cannot rely on diffusion alone as many large organelles and macromolecular complexes are essentially immobilized by the dense meshwork of the cytosol. One strategy used by cells to overcome diffusion is to harness the free energy liberated by ATP hydrolysis through molecular motors. Myosins are a family of actin based molecular motors that have evolved a variety of ways to contribute to cellular organization through numerous modifications to the manner they convert that free energy into mechanical work.
Collapse
|
36
|
Izagirre N, García I, Junquera C, de la Rúa C, Alonso S. A scan for signatures of positive selection in candidate loci for skin pigmentation in humans. Mol Biol Evol 2006; 23:1697-706. [PMID: 16757656 DOI: 10.1093/molbev/msl030] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although the combination of pale skin and intense sun exposure results in an important health risk for the individual, it is less clear if at the population level this risk has possessed an evolutionary meaning. In this sense, a number of adaptive hypotheses have been put forward to explain the evolution of human skin pigmentation, such as photoprotection against sun-induced cancer, sexual selection, vitamin D synthesis or photoprotection of photolabile compounds, among others. It is expected that if skin pigmentation is adaptive, we might be able to see the signature of positive selection on some of the genes involved. In order to detect this signature, we analyze a battery of 81 candidate loci by means of phylogenetic and population genetic tests. Our results indicate that both light and dark skin may possess adaptive value. Of the main loci presenting this signature, TP53BP1 shows clear evidence of adaptive selection in Africans, whereas TYRP1 and SLC24A5 show evidence of adaptive selection in Caucasians. Although we cannot offer a mechanism that based on these genes explains the advantage of light skin, if TP53BP1, and perhaps RAD50, have truly conferred an adaptive value to the African population analyzed, photoprotection against sun-induced skin damage/cancer might be proposed as a mechanism that has driven the evolution of human skin pigmentation.
Collapse
Affiliation(s)
- Neskuts Izagirre
- Department Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country, Leioa, Bizkaia, Spain
| | | | | | | | | |
Collapse
|
37
|
Reiners J, Nagel-Wolfrum K, Jürgens K, Märker T, Wolfrum U. Molecular basis of human Usher syndrome: deciphering the meshes of the Usher protein network provides insights into the pathomechanisms of the Usher disease. Exp Eye Res 2006; 83:97-119. [PMID: 16545802 DOI: 10.1016/j.exer.2005.11.010] [Citation(s) in RCA: 197] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2005] [Revised: 11/15/2005] [Accepted: 11/21/2005] [Indexed: 11/17/2022]
Abstract
Usher syndrome (USH) is the most frequent cause of combined deaf-blindness in man. It is clinically and genetically heterogeneous and at least 12 chromosomal loci are assigned to three clinical USH types, namely USH1A-G, USH2A-C, USH3A (Davenport, S.L.H., Omenn, G.S., 1977. The heterogeneity of Usher syndrome. Vth Int. Conf. Birth Defects, Montreal; Petit, C., 2001. Usher syndrome: from genetics to pathogenesis. Annu. Rev. Genomics Hum. Genet. 2, 271-297). Mutations in USH type 1 genes cause the most severe form of USH. In USH1 patients, congenital deafness is combined with a pre-pubertal onset of retinitis pigmentosa (RP) and severe vestibular dysfunctions. Those with USH2 have moderate to severe congenital hearing loss, non-vestibular dysfunction and a later onset of RP. USH3 is characterized by variable RP and vestibular dysfunction combined with progressive hearing loss. The gene products of eight identified USH genes belong to different protein classes and families. There are five known USH1 molecules: the molecular motor myosin VIIa (USH1B); the two cell-cell adhesion cadherin proteins, cadherin 23 (USH1D) and protocadherin 15, (USH1F) and the scaffold proteins, harmonin (USH1C) and SANS (USH1G). In addition, two USH2 genes and one USH3A gene have been identified. The two USH2 genes code for the transmembrane protein USH2A, also termed USH2A ("usherin") and the G-protein-coupled 7-transmembrane receptor VLGR1b (USH2C), respectively, whereas the USH3A gene encodes clarin-1, a member of the clarin family which exhibits 4-transmembrane domains. Molecular analysis of USH1 protein function revealed that all five USH1 proteins are integrated into a protein network via binding to PDZ domains in the USH1C protein harmonin. Furthermore, this scaffold function of harmonin is supported by the USH1G protein SANS. Recently, we have shown that the USH2 proteins USH2A and VLGR1b as well as the candidate for USH2B, the sodium bicarbonate co-transporter NBC3, are also integrated into this USH protein network. In the inner ear, these interactions are essential for the differentiation of hair cell stereocilia but may also participate in the mechano-electrical signal transduction and the synaptic function of maturated hair cells. In the retina, the co-expression of all USH1 and USH2 proteins at the synapse of photoreceptor cells indicates that they are organized in an USH protein network there. The identification of the USH protein network indicates a common pathophysiological pathway in USH. Dysfunction or absence of any of the molecules in the mutual "interactome" related to the USH disease may lead to disruption of the network causing senso-neuronal degeneration in the inner ear and the retina, the clinical symptoms of USH.
Collapse
Affiliation(s)
- Jan Reiners
- Institute of Zoology, Department of Cell and Matrix Biology, Johannes Gutenberg University of Mainz, Müllerweg 6, D-55099 Mainz, Germany
| | | | | | | | | |
Collapse
|
38
|
Watanabe S, Ikebe R, Ikebe M. Drosophila myosin VIIA is a high duty ratio motor with a unique kinetic mechanism. J Biol Chem 2006; 281:7151-60. [PMID: 16415346 DOI: 10.1074/jbc.m511592200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mutations of myosin VIIA cause deafness in various species from human and mice to Zebrafish and Drosophila. We analyzed the kinetic mechanism of the ATPase cycle of Drosophila myosin VIIA by using a single-headed construct with the entire neck domain. The steady-state ATPase activity (0.06 s(-1)) was markedly activated by actin to yield V(max) and K(ATPase) of 1.72 s(-1) and 3.2 microm, respectively. The most intriguing finding is that the ATP hydrolysis predominantly takes place in the actin-bound form (actin-attached hydrolysis) for the actomyosin VIIA ATPase reaction. The ATP hydrolysis rate was much faster for the actin-attached form than the dissociated form, in contrast to other myosins reported so far. Both the ATP hydrolysis step and the phosphate release step were significantly faster than the entire ATPase cycle rate, thus not rate-determining. The rate of ADP dissociation from actomyosin VIIA was 1.86 s(-1), which was comparable with the overall ATPase cycle rate, thus assigned to be a rate-determining step. The results suggest that Drosophila myosin VIIA spends the majority of the ATPase cycle in an actomyosin.ADP form, a strong actin binding state. The duty ratio calculated from our kinetic model was approximately 0.9. Therefore, myosin VIIA is classified to be a high duty ratio motor. The present results suggested that myosin VIIA can be a processive motor to serve cargo trafficking in cells once it forms a dimer structure.
Collapse
Affiliation(s)
- Shinya Watanabe
- Department of Physiology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, USA
| | | | | |
Collapse
|